Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 188
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 970: 176461, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38460658

ABSTRACT

Our previous studies have showed that sulfatide-reactive type II NKT (i.e. variant NKT, vNKT) cells inhibit the immunogenic maturation during the development of mature lung dendritic cells (LDCs), leading todeclined allergic airway inflammation in asthma. Nonetheless, the specific immunoregulatory roles of vNKT cells in LDC-mediated Th2 cell responses remain incompletely understood. Herein, we found that administration of sulfatide facilitated the generation of CD4+FoxP3+ regulatory T (Treg) cells in the lungs of wild-type mice, but not in CD1d-/- and Jα18-/- mice, after ovalbumin or house dust mite exposure. This finding implies that the enhancement of lung Treg cells by sulfatide requires vNKT cells, which dependent on invariant NKT (iNKT) cells. Furthermore, the CD4+FoxP3+ Treg cells induced by sulfatide-reactive vNKT cells were found to be associated with PD-L1 molecules expressed on LDCs, and this association was dependent on iNKT cells. Collectively, our findings suggest that in asthma-mimicking murine models, sulfatide-reactive vNKT cells facilitate the generation of lung Treg cells through inducing tolerogenic properties in LDCs, and this process is dependent on the presence of lung iNKT cells. These results may provide a potential therapeutic approach to treat allergic asthma.


Subject(s)
Asthma , T-Lymphocytes, Regulatory , Mice , Animals , Sulfoglycosphingolipids/pharmacology , Sulfoglycosphingolipids/metabolism , Sulfoglycosphingolipids/therapeutic use , Mice, Inbred BALB C , Lung , Asthma/drug therapy , Inflammation/metabolism , Dendritic Cells , Forkhead Transcription Factors/metabolism , Disease Models, Animal
2.
Ann Clin Transl Neurol ; 11(2): 328-341, 2024 02.
Article in English | MEDLINE | ID: mdl-38146590

ABSTRACT

OBJECTIVE: To evaluate the longitudinal correlations between sulfatide/lysosulfatide levels and central and peripheral nervous system function in children with metachromatic leukodystrophy (MLD) and to explore the impact of intravenous recombinant human arylsulfatase A (rhASA) treatment on myelin turnover. METHODS: A Phase 1/2 study of intravenous rhASA investigated cerebrospinal fluid (CSF) and sural nerve sulfatide levels, 88-item Gross Motor Function Measure (GMFM-88) total score, sensory and motor nerve conduction, brain N-acetylaspartate (NAA) levels, and sural nerve histology in 13 children with MLD. Myelinated and unmyelinated nerves from an untreated MLD mouse model were also analyzed. RESULTS: CSF sulfatide levels correlated with neither Z-scores for GMFM-88 nor brain NAA levels; however, CSF sulfatide levels correlated negatively with Z-scores of nerve conduction parameters, number of large (≥7 µm) myelinated fibers, and myelin/fiber diameter slope, and positively with nerve g-ratios and cortical latencies of somatosensory-evoked potentials. Quantity of endoneural litter positively correlated with sural nerve sulfatide/lysosulfatide levels. CSF sulfatide levels decreased with continuous high-dose treatment; this change correlated with improved nerve conduction. At 26 weeks after treatment, nerve g-ratio decreased by 2%, and inclusion bodies per Schwann cell unit increased by 55%. In mice, abnormal sulfatide storage was observed in non-myelinating Schwann cells in Remak bundles of sciatic nerves but not in unmyelinated urethral nerves. INTERPRETATION: Lower sulfatide levels in the CSF and peripheral nerves correlate with better peripheral nerve function in children with MLD; intravenous rhASA treatment may reduce CSF sulfatide levels and enhance sulfatide/lysosulfatide processing and remyelination in peripheral nerves.


Subject(s)
Leukodystrophy, Metachromatic , Psychosine/analogs & derivatives , Child , Humans , Mice , Animals , Leukodystrophy, Metachromatic/drug therapy , Sulfoglycosphingolipids/pharmacology , Cerebroside-Sulfatase , Sciatic Nerve/pathology
3.
Diabetes Obes Metab ; 25(9): 2514-2525, 2023 09.
Article in English | MEDLINE | ID: mdl-37246802

ABSTRACT

AIM: To study the effect of sulfatide on gene expression and proliferation of human primary fibroblasts induced by insulin, insulin-like growth factor-1 and human growth hormone. MATERIALS AND METHODS: Human primary fibroblasts were exposed to 1, 3 and 30 µM of sulfatide or its precursor galactosylceramide (GalCer). Proliferation was determined by 3 H-thymidine incorporation and gene expression via microarray analysis. RESULTS: Sulfatide and GalCer reduced the growth rate of fibroblasts by 32%-82% when exposed to 0.5 nM insulin. After challenge with 120 µM of H2 O2 , sulfatide reduced membrane leakage. Fibroblast gene expression was altered by sulfatide in gene pathways associated with cell cycle/growth, transforming growth factor-ß function, and encoding of proteins involved in intracellular signalling. NFKBIA, a key control element in NF-кB regulation, was decreased 2-fold by sulfatide. CONCLUSIONS: Sulfatide strongly inhibits fibroblast growth. We therefore suggest the addition of sulfatide to injectable commercial insulin formulations, which would reduce adverse fibroblast growth and improve well-being in patients with diabetes.


Subject(s)
Insulin , Sulfoglycosphingolipids , Humans , Insulin/pharmacology , Insulin/metabolism , Sulfoglycosphingolipids/metabolism , Sulfoglycosphingolipids/pharmacology , Insulin, Regular, Human , Fibroblasts/metabolism , Oxidative Stress
4.
Int J Biochem Cell Biol ; 159: 106419, 2023 06.
Article in English | MEDLINE | ID: mdl-37086817

ABSTRACT

Iron chelators, such as deferoxamine, exert an anticancer effect by altering the activity of biomolecules critical for regulation of the cell cycle, cell metabolism, and apoptotic processes. Thus, iron chelators are sometimes used in combination with radio- and/or chemotherapy in the treatment of cancer. The possibility that deferoxamine could induce a program of senescence similar to radio- and/or chemotherapy, fostering adaptation in the treatment of cancer cells, is not fully understood. Using established biochemical techniques, biomarkers linked to lipid composition, and coherent anti-Stokes Raman scattering microscopy, we demonstrated that hepatocellular carcinoma-derived HepG2 cells survive after deferoxamine treatment, acquiring phenotypic traits and representative hallmarks of senescent cells. The results support the view that deferoxamine acts in HepG2 cells to produce oxidative stress-induced senescence by triggering sequential mitochondrial and lysosomal dysfunction accompanied by autophagy blockade. We also focused on the lipidome of senescent cells after deferoxamine treatment. Using mass spectrometry, we found that the deferoxamine-induced senescent cells presented marked remodeling of the phosphoinositol, sulfatide, and cardiolipin profiles, which all play a central role in cell signaling cascades, intracellular membrane trafficking, and mitochondria functions. Detection of alterations in glycosphingolipid sulfate species suggested modifications in ceramide generation, and turnover is frequently described in cancer cell survival and resistance to chemotherapy. Blockade of ceramide generation may explain autophagic default, resistance to apoptosis, and the onset of senescence.


Subject(s)
Deferoxamine , Sulfoglycosphingolipids , Humans , Deferoxamine/pharmacology , Deferoxamine/metabolism , Sulfoglycosphingolipids/metabolism , Sulfoglycosphingolipids/pharmacology , Hep G2 Cells , Iron Chelating Agents/pharmacology , Iron Chelating Agents/metabolism , Mitochondria/metabolism , Cellular Senescence
5.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34290146

ABSTRACT

Many endogenous molecules, mostly proteins, purportedly activate the Toll-like receptor 4 (TLR4)-myeloid differentiation factor-2 (MD-2) complex, the innate immune receptor for lipopolysaccharide (LPS) derived from gram-negative bacteria. However, there is no structural evidence supporting direct TLR4-MD-2 activation by endogenous ligands. Sulfatides (3-O-sulfogalactosylceramides) are natural, abundant sulfated glycolipids that have variously been shown to initiate or suppress inflammatory responses. We show here that short fatty acid (FA) chain sulfatides directly activate mouse TLR4-MD-2 independent of CD14, trigger MyD88- and TRIF-dependent signaling, and stimulate tumor necrosis factor α (TNFα) and type I interferon (IFN) production in mouse macrophages. In contrast to the agonist activity toward the mouse receptor, the tested sulfatides antagonize TLR4-MD-2 activation by LPS in human macrophage-like cells. The agonistic and antagonistic activities of sulfatides require the presence of the sulfate group and are inversely related to the FA chain length. The crystal structure of mouse TLR4-MD-2 in complex with C16-sulfatide revealed that three C16-sulfatide molecules bound to the MD-2 hydrophobic pocket and induced an active dimer conformation of the receptor complex similar to that induced by LPS or lipid A. The three C16-sulfatide molecules partially mimicked the detailed interactions of lipid A to achieve receptor activation. Our results suggest that sulfatides may mediate sterile inflammation or suppress LPS-stimulated inflammation, and that additional endogenous negatively charged lipids with up to six lipid chains of limited length might also bind to TLR4-MD-2 and activate or inhibit this complex.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Lymphocyte Antigen 96/metabolism , Myeloid Differentiation Factor 88/metabolism , Sulfoglycosphingolipids/pharmacology , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cell Line , Female , Humans , Lymphocyte Antigen 96/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Dynamics Simulation , Myeloid Differentiation Factor 88/genetics , Sulfoglycosphingolipids/chemistry , Toll-Like Receptor 4/genetics
6.
Int J Biol Macromol ; 147: 792-798, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-31739035

ABSTRACT

Sulfatide is associated with numerous health problems, affecting different parts of the human body, including the metastasis; however, the underlying mechanisms are yet to be fully elucidated. Sulfatide has been used to potential inhibitor for tumor cell metastasis. In the present study we synthesized oleic acid sulfated chitosan (OlcShCs). It shows structural similarity to sulfatide because of its functional groups (sulfate and fatty acyl chains). Chitosan has smart properties such as biocompatibility, biodegradability and non-toxicity. We have prepared oleic acid sulfated chitosan (OlcShCs) by chitosan modification to mimic sulfatide. Its structure was characterized by FT-IR, H-NMR, and thermogravimetric analysis. After characterization studies its antimicrobial, antifungal and cytotoxic properties were investigated. Oleic acid sulfated chitosan (OlcShCs) was tested for its anti-cancer potential against human cancer cell lines (HeLa (ATCC® CCL-2™)) for 24 h, 48 h and 72 h using the MTT assays. This new material which is soluble at physiological conditions, is a potential candidate for further metastasis inhibition investigations.


Subject(s)
Antineoplastic Agents , Chitosan , Neoplasms/drug therapy , Oleic Acid , Sulfoglycosphingolipids , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , HeLa Cells , Humans , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Oleic Acid/chemistry , Oleic Acid/pharmacology , Sulfoglycosphingolipids/chemical synthesis , Sulfoglycosphingolipids/chemistry , Sulfoglycosphingolipids/pharmacology
7.
Am J Physiol Lung Cell Mol Physiol ; 317(5): L578-L590, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31432714

ABSTRACT

Our previous study showed that sulfatide-activated type II natural killer T (NKT) cells can prevent allergic airway inflammation in an ovalbumin (OVA)-induced murine model of asthma, but the underlying mechanism is unclear. Recently, sulfatide-activated type II NKT cells were shown to modulate the function of dendritic cells in experimental autoimmune encephalomyelitis and nonobese diabetic mice. Thus, it was hypothesized that sulfatide-activated type II NKT cells may modulate the function of lung dendritic cells (LDCs) in asthmatic mice. Our data showed that, in our mouse models, activation of type II NKT cells by sulfatide administration and adoptive transfer of sulfatide-activated type II NKT cells resulted in reduced expression of surface maturation markers and proinflammatory cytokine production of LDCs. LDCs from sulfatide-treated asthmatic mice, in contrast to LDCs from PBS-treated asthmatic mice, significantly reduced allergic airway inflammation in vivo. However, we found no influence of sulfatide-activated type II NKT cells on the phenotypic and functional maturation of bone marrow-derived dendritic cells in vitro. In addition, adoptive transfer of sulfatide-activated type II NKT cells did not influence the phenotypic and functional maturation of LDCs in CD1d-/- mice, which lack both type I and II NKT cells, immunized and challenged with OVA. Our data reveal that sulfatide-activated type II NKT cells can suppress immunogenic maturation of LDCs to reduce allergic airway inflammation in mouse models of asthma, and it is possible that the immunomodulatory effect needs type I NKT cells.


Subject(s)
Asthma/immunology , Dendritic Cells/immunology , Inflammation Mediators/metabolism , Lung/immunology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Sulfoglycosphingolipids/pharmacology , Animals , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Lung/drug effects , Lung/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/metabolism , Ovalbumin/administration & dosage
8.
J Neuroimmunol ; 326: 55-61, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30481614

ABSTRACT

Sulfatides have immunomodulatory functions, and play protective roles in multiple autoimmune diseases. In the present study, we showed that sulfatides ameliorated experimental autoimmune neuritis in Lewis rats induced with bovine peripheral myelin, which was associated with decreased proportions of Th1 and Th17 cells. Furthermore, compared control group, cells from sulfatide-treated rats exhibited lower potential in proliferation and IL-17 secretion in the presence of BPM or ConA in vitro. Moreover, sulfatides also reduced the proportions of NK and NKT cells. In summary, our study indicated that sulfatides might become a new therapeutic agent in Guillain-Barré syndrome in the future.


Subject(s)
Neuritis, Autoimmune, Experimental/immunology , Sulfoglycosphingolipids/pharmacology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Female , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Rats , Rats, Inbred Lew , Th1 Cells/drug effects , Th17 Cells/drug effects
9.
J Mol Cell Biol ; 11(5): 421-432, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30215728

ABSTRACT

Paired amphipathic helix protein (SIN3B) is a transcription corepressor for many genes. Here we show a different regulation mechanism of integrin αV gene expression by SIN3B in human hepatocellular carcinoma (HCC). We first observed a close relationship between Integrin αV and SIN3B expressions in HCC patients and tumor cell lines with different metastatic potentials. Overexpression of SIN3B significantly accelerated the cell migration rate of SMMC-7721, but failed when integrin αV expression was silenced. Interestingly, SIN3B stimulated integrin αV subunit promoter activity only in the presence of sulfatide. Importantly, SIN3B was identified in the complex with sulfatide by mass spectrometry. Fat blot assay indicated that SIN3B specifically interacted with sulfatide. Molecular modeling suggested that sulfatide induced the conformational change of SIN3B from compacted α-helices to a relaxed ß-sheet in PAH2 domain. The data of immunoprecipitation and ChIP assay indicated that altered SIN3B lost the binding affinity with MAD1 and HDAC2, which reduced the recruitment of HDAC2 on integrin αV gene promoter and prevented the deacetylation of the histone 3. In conclusion, this study demonstrated that SIN3B promoted the transcriptional activation of the integrin αV subunit gene promoter by reducing interaction with HDAC2.


Subject(s)
Carcinoma, Hepatocellular/pathology , Integrin alphaV/metabolism , Liver Neoplasms/pathology , Repressor Proteins/metabolism , Acetylation , Carcinoma, Hepatocellular/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Histone Deacetylase 2/chemistry , Histone Deacetylase 2/metabolism , Histones/metabolism , Humans , Integrin alphaV/genetics , Liver Neoplasms/metabolism , Molecular Dynamics Simulation , Promoter Regions, Genetic , Protein Binding/drug effects , Protein Structure, Secondary , Protein Subunits/genetics , Protein Subunits/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Sulfoglycosphingolipids/pharmacology , Transcription, Genetic/drug effects
10.
Oncotarget ; 7(24): 36563-36576, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27145276

ABSTRACT

Integrin αVß3 is a malignant driver of anchorage-independence and tumor angiogenesis, but its dysregulation in hepatocellular carcinoma (HCC) remains unclear. In this study, we observed that sulfatide significantly promoted integrin αV(ITGAV) expression and wound closure in HCC. We also noted that elevated sulfatide profoundly stimulated integrin αVß3 clustering and signaling. In the cells with integrin αVß3 clustering induced by sulfatide, integrin ß3 subunit was phosphorylated. Simultaneously, focal adhesion kinase (FAK), Src and paxillin were also phosphorylated. Treatment with FAK inhibitor resulted in robust suppression of FAK-Y397 and Src-Y416 phosphorylation stimulated by sulfatide, but not suppression of integrin ß3 phosphorylation. Src inhibitors repressed Src-Y416 and FAK Y861 and Y925 phosphorylation, but not FAK-Y397 and integrin ß3 phosphorylation. After mutation of integrin ß3 (Y773F and Y785F), FAK or Src phosphorylation failed to be stimulated by sulfatide. Moreover, ß3 Y773 and Y785 phosphorylation was suppressed by insulin-like growth factor receptor knockdown even in cells stimulated by sulfatide. In assays of immunoprecipitation and immunostaining with integrin αV or ß3 antibody, labeled sulfatide was found in the complex and co-localized with integrin αVß3. Taken together, this study demonstrated that elevated sulfatide bound to integrin αVß3 and induced clustering and phosphorylation of αVß3 instead of matrix ligand binding, triggering outside-in signaling.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Integrin alphaVbeta3/genetics , Signal Transduction/drug effects , Sulfoglycosphingolipids/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin alphaVbeta3/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Paxillin/metabolism , Phosphorylation/drug effects , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Sulfoglycosphingolipids/metabolism , Tyrosine/genetics , Tyrosine/metabolism , src-Family Kinases/metabolism
11.
Neurochem Res ; 41(1-2): 130-43, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26542149

ABSTRACT

Sulfatide (3-O-sulfogalactosylceramide, SM4s) was isolated by Thudichum from the human brain in 1884. Together with galactosylceramide, its direct metabolic precursor in the biosynthetic pathway, sulfatide is highly enriched in myelin in the central and peripheral nervous system, and it has been implicated in several aspects of the biology of myelin-forming cells. Studies obtained using galactolipid-deficient mice strongly support the notion that sulfatide plays critical roles in the correct structure and function of myelin membrane. A number of papers are suggesting that these roles are mediated by a specific function of sulfatide in the lateral organization of myelin membrane, thus affecting the sorting, lateral assembly, membrane dynamics and also the function of specific myelin proteins in different substructures of the myelin sheath. The consequences of altered sulfatide metabolism and sulfatide-mediated myelin organization with respect to myelin diseases are still poorly understood, but it's very likely that sulfatide might represent not only a critical player in the pathogenesis of several diseases, including multiple sclerosis and Alzheimer's disease, but also a potentially promising therapeutic target.


Subject(s)
Myelin Sheath/metabolism , Sulfoglycosphingolipids/pharmacology , Animals , Antibodies/immunology , Humans , Mice , Sulfoglycosphingolipids/immunology
12.
Mol Immunol ; 66(2): 463-71, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26021803

ABSTRACT

Cholera toxin (CTX) is a virulent factor of Vibrio cholerae that causes life-threatening diarrheal disease. Its non-toxic subunit CTB has been extensively studied for vaccine delivery. In immune cells, CTB induces a number of signaling molecules related to cellular activation and cytokine production. The mechanisms by which CTB exerts its immunological effects are not understood. We report here the immunological targets of CTB. The unexpected finding that GM1 ganglioside inhibited NF-κB activation in human monocytes stimulated with CTX and agonists of Toll-like receptors (TLR) suggests the possibility of CTX-TLR interaction. Indeed, CTX-induced IL-6 production was substantially reduced in MyD88(-/-) or TLR4(-/-) macrophages. Ectopic expression of TLR4 was required for CTX-induced NF-κB activation in HEK 293 cells. Furthermore, the inflammatory capacity of CTB was lost in the absence of TLR4, adaptor protein FcRγ, or its downstream signaling molecule CARD9. Attempts have been made to identify CTB-binding targets from various C-type lectin and immunoglobulin-like receptors. CTB targeted not only GM1 and TLR4 but also TREM2 and LMIR5/CD300b. CTB-TREM2 interaction initiated signal transduction through adaptor protein DAP12. The binding of CTB inhibited LMIR5 activation induced by its endogenous ligand 3-O-sulfo-ß-d-galactosylceramide C24:1. In summary, CTB targets TLR4, FcRγ-CARD9, TREM2, and LMIR5. These findings provide new insights into the immunobiology of cholera toxin.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Cholera Toxin/pharmacology , Macrophages, Peritoneal/drug effects , Membrane Glycoproteins/immunology , Receptors, IgG/immunology , Receptors, Immunologic/immunology , Toll-Like Receptor 4/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Binding Sites , CARD Signaling Adaptor Proteins/genetics , G(M1) Ganglioside/immunology , G(M1) Ganglioside/pharmacology , Gene Expression Regulation , HEK293 Cells , Humans , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Primary Cell Culture , Protein Binding , Receptors, IgG/genetics , Receptors, Immunologic/genetics , Signal Transduction , Sulfoglycosphingolipids/pharmacology , Toll-Like Receptor 4/genetics
13.
Hepatology ; 61(4): 1357-69, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25477000

ABSTRACT

UNLABELLED: Innate immune mechanisms leading to liver injury subsequent to chronic alcohol ingestion are poorly understood. Natural killer T (NKT) cells, enriched in the liver and comprised of at least two distinct subsets, type I and II, recognize different lipid antigens presented by CD1d molecules. We have investigated whether differential activation of NKT cell subsets orchestrates inflammatory events leading to alcoholic liver disease (ALD). We found that after chronic plus binge feeding of Lieber-DeCarli liquid diet in male C57BL/6 mice, type I, but not type II, NKT cells are activated, leading to recruitment of inflammatory Gr-1(high) CD11b(+) cells into the liver. A central finding is that liver injury after alcohol feeding is dependent upon type I NKT cells. Thus, liver injury is significantly inhibited in Jα18(-/-) mice deficient in type I NKT cells as well as after their inactivation by sulfatide-mediated activation of type II NKT cells. Furthermore, we have identified a novel pathway involving all-trans retinoic acid (ATRA) and its receptor (RARγ) signaling that inhibits type I NKT cells and, consequently, ALD. A semiquantitative polymerase chain reaction analysis of hepatic gene expression of some of the key proinflammatory molecules shared in human disease indicated that their up-regulation in ALD is dependent upon type I NKT cells. CONCLUSIONS: Type I, but not type II, NKT cells become activated after alcohol feeding. Type I NKT cell-induced inflammation and neutrophil recruitment results in liver tissue damage whereas type II NKT cells protect from injury in ALD. Inhibition of type I NKT cells by retinoids or by sulfatide prevents ALD. Given that the CD1d pathway is highly conserved between mice and humans, NKT cell subsets might be targeted for potential therapeutic intervention in ALD.


Subject(s)
Liver Diseases, Alcoholic/immunology , Liver Diseases, Alcoholic/prevention & control , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/physiology , Retinoids/pharmacology , Retinoids/therapeutic use , Sulfoglycosphingolipids/pharmacology , Sulfoglycosphingolipids/therapeutic use , Animals , Male , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/classification
14.
Int J Nanomedicine ; 9: 3971-85, 2014.
Article in English | MEDLINE | ID: mdl-25170267

ABSTRACT

Targeted nanoparticle (NP) delivery vehicles are emerging technologies, the full potential of which has yet to be realized. Sulfatide is known to bind to extracellular matrix glycoproteins that are highly expressed in breast tumors. In this study, we report for the first time the combination of sulfatide and lipid perfluorooctylbromide NPs as a targeted breast cancer delivery vehicle for paclitaxel (PTX). PTX-sulfatide-containing lipid perfluorooctylbromide NPs (PTX-SNPs) were prepared using the emulsion/solvent evaporation method. PTX-SNPs exhibited a spherical shape, small particle size, high encapsulation efficiency, and a biphasic release in phosphate-buffered solution. The cytotoxicity study and cell apoptosis assay revealed that blank sulfatide-containing lipid perfluorooctylbromide NPs (SNPs) had no cytotoxicity, whereas PTX-SNPs had greater EMT6 cytotoxicity levels than PTX-lipid perfluorooctylbromide NPs (PTX-NPs) and free PTX. An in vitro cellular uptake study revealed that SNPs can deliver greater amounts of drug with more efficient and immediate access to intracellular targets. In vivo biodistribution measured using high-performance liquid chromatography confirmed that the PTX-SNPs can target breast tumor tissues to increase the accumulation of PTX in these tissues. The in vivo tumor inhibition ability of PTX-SNPs was remarkably higher than PTX-NPs and free PTX. Furthermore, toxicity studies suggested that the blank SNPs had no systemic toxicity. All results suggested that SNPs may serve as efficient PTX delivery vehicles targeting breast carcinoma.


Subject(s)
Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Mammary Neoplasms, Experimental/metabolism , Nanoparticles/chemistry , Paclitaxel/chemistry , Sulfoglycosphingolipids/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/pharmacology , Female , Fluorocarbons/chemistry , Fluorocarbons/pharmacology , Hydrocarbons, Brominated , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Paclitaxel/pharmacology , Sulfoglycosphingolipids/pharmacology , Tissue Distribution
15.
Glia ; 62(6): 927-42, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24578319

ABSTRACT

In the central nervous system, the extracellular matrix (ECM) compound laminin-2, present on developing axons, is essential in regulating oligodendrocyte (OLG) maturation. For example, laminin-2 is involved in mediating interactions between integrins and growth factors, initially localizing in separate membrane microdomains. The galactosphingolipid sulfatide is an important constituent of these microdomains and may serve as a receptor for laminin-2. Here, we investigated whether sulfatide interferes with ECM-integrin interactions and, in this manner, modulates OLG maturation. Our data reveal that disruption of laminin-2-sulfatide interactions impeded OLG differentiation and myelin-like membrane formation. On laminin-2, but not on (re)myelination-inhibiting fibronectin, sulfatide laterally associated with integrin α6 in membrane microdomains. Sulfatide was partly excluded from membrane microdomains on fibronectin, thereby likely precluding laminin-2-mediated myelination. Anti-sulfatide antibodies disrupted integrin α6-PDGFαR interactions on laminin-2 and induced demyelination in myelinated spheroid cultures, but intriguingly stimulated myelin-like membrane formation on fibronectin. Taken together, these findings highlight the importance of laminin-sulfatide interactions in the formation of functional membrane microdomains essential for myelination. Thus, laminin-sulfatide interactions might control the asynchronous localized differentiation of OLGs, thereby allowing myelination to be triggered by axonal demand. Given the accumulation of fibronectin in multiple sclerosis lesions, the findings also provide a molecular rationale for the potential of anti-sulfatide antibodies to trigger quiescent endogenous OLG progenitor cells in axon remyelination. GLIA 2014;62:927-942.


Subject(s)
Cell Enlargement , Cell Proliferation/physiology , Extracellular Matrix/physiology , Sulfoglycosphingolipids/pharmacology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Enlargement/drug effects , Cell Line , Extracellular Matrix/drug effects , Laminin/physiology , Nerve Fibers, Myelinated/physiology , Oligodendroglia , Rats , Rats, Wistar
16.
PLoS One ; 8(4): e61092, 2013.
Article in English | MEDLINE | ID: mdl-23593400

ABSTRACT

Influenza A virus (IAV) generally causes caspase-dependent apoptosis based on caspase-3 activation, resulting in nuclear export of newly synthesized viral nucleoprotein (NP) and elevated virus replication. Sulfatide, a sulfated galactosylsphingolipid, enhances IAV replication through promoting newly synthesized viral NP export induced by association of sulfatide with hemagglutinin delivered to the cell surface. Here, we demonstrated that sulfatide is involved in caspase-3-independent apoptosis initiated by the PB1-F2 protein of IAV by using genetically sulfatide-produced cells and PB1-F2-deficient IAVs. Sulfatide-deficient COS7 cells showed no virus-induced apoptosis, whereas SulCOS1 cells, sulfatide-enriched COS7 cells that genetically expressed the two transferases required for sulfatide synthesis from ceramide, showed an increase in IAV replication and were susceptible to caspase-3-independent apoptosis. Additionally, PB1-F2-deficient IAVs, which were generated by using a plasmid-based reverse genetics system from a genetic background of A/WSN/33 (H1N1), demonstrated that PB1-F2 contributed to caspase-3-independent apoptosis in IAV-infected SulCOS1 cells. Our results show that sulfatide plays a critical role in efficient IAV propagation via caspase-3-independent apoptosis initiated by the PB1-F2 protein.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/metabolism , Influenza A virus/metabolism , Sulfoglycosphingolipids/pharmacology , Viral Proteins/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Protein Transport , RNA Interference , Virus Replication
17.
J Lipid Res ; 54(4): 936-52, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23345412

ABSTRACT

Integrin is important in migration and metastasis of tumor cells. Changes of integrin expression and distribution will cause an alteration of cellular adhesion and migration behaviors. In this study, we investigated sulfatide regulation of the integrin αV subunit expression in hepatoma cells and observed that either exogenous or endogenous sulfatide elicited a robust upregulation of integrin αV subunit mRNA and protein expression in hepatoma cells. This regulatory effect occurred with a corresponding phosphorylation (T739) of the transcription factor Sp1. Based on the electrophoretic mobility shift assay, sulfatide enhanced the integrin αV promoter activity and strengthened the Sp1 complex super-shift. The results of chromatin immunoprecipitation analysis also indicated that sulfatide enhanced Sp1 binding to the integrin αV promoter in vivo. Silence of Sp1 diminished the stimulation of integrin αV expression by sulfatide. In the early stage of sulfatide stimulation, phosphorylation of Erk as well as c-Src was noted, and inhibition of Erk activation with either U0126 or PD98059 significantly suppressed Sp1 phosphorylation and integrin αV expression. We demonstrated that sulfatide regulated integrin αV expression and cell adhesion, which was associated with Erk activation.


Subject(s)
Integrin alphaV/metabolism , Sulfoglycosphingolipids/pharmacology , Butadienes/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cerebrosides/metabolism , Chromatin Immunoprecipitation , Flavonoids/pharmacology , Humans , Nitriles/pharmacology , Phosphorylation/drug effects , Sp1 Transcription Factor/metabolism
18.
Glia ; 61(4): 466-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23322453

ABSTRACT

Cerebroside sulfotransferase (CST) catalyzes the production of sulfatide, which is one of the major glycolipids in myelin. Homozygous CST knockout mice were shown to be completely deficient in sulfatide. They were born healthy but began to display progressive neurological deficits from 6 weeks of age. Severe abnormalities of paranodal regions and changes in axonal ion channel distribution were prominent in both the central and peripheral nervous systems. But whether partial decreases in myelin sulfatide levels influence paranodal formation, as well as nerve conduction velocity (NCV), is largely unknown. To determine the functional significance of sulfatide content in myelin, we performed electrophysiological, morphological, and biochemical analyses using heterozygote, homozygote, and wild-type mouse peripheral nerves and compared the results with individual sulfatide content. NCVs were significantly reduced in homozygote animals compared with wild-type mice. In contrast, these values were markedly varied in individual heterozygote mice. On the basis of NCV values, we divided heterozygous mice into two groups: mice with mild but significant reduction of NCV and those with normal NCV. Teased nerve fibers obtained from individual mouse sciatic nerves were immunostained, and Na(+) channel and Caspr cluster lengths were measured to determine abnormal levels of junctional formation at the paranode. Furthermore, sulfatide content in each sciatic nerve was examined by thin layer chromatography. The results demonstrated significant correlations among sulfatide level, severity of paranodal abnormality, and reduction of NCV. Thus, the fine regulation of myelin sulfatide content by CST is important for normal function of myelinated axons.


Subject(s)
Axons/metabolism , Axons/physiology , Myelin Sheath/metabolism , Neural Conduction/physiology , Neuroglia/metabolism , Neuromuscular Junction/physiology , Sciatic Nerve/physiology , Sulfoglycosphingolipids/pharmacology , Animals , Axons/chemistry , Axons/enzymology , Genetic Carrier Screening , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/genetics , Neuroglia/chemistry , Neuroglia/enzymology , Sciatic Nerve/chemistry , Sciatic Nerve/enzymology , Sulfotransferases/deficiency , Sulfotransferases/genetics
19.
Cancer Res ; 73(5): 1514-23, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23319803

ABSTRACT

The nature of the regulatory cell types that dominate in any given tumor is not understood at present. Here, we addressed this question for regulatory T cells (Treg) and type II natural killer T (NKT) cells in syngeneic models of colorectal and renal cancer. In mice with both type I and II NKT cells, or in mice with neither type of NKT cell, Treg depletion was sufficient to protect against tumor outgrowth. Surprisingly, in mice lacking only type I NKT cells, Treg blockade was insufficient for protection. Thus, we hypothesized that type II NKT cells may be neutralized by type I NKT cells, leaving Tregs as the primary suppressor, whereas in mice lacking type I NKT cells, unopposed type II NKT cells could suppress tumor immunity even when Tregs were blocked. We confirmed this hypothesis in 3 ways by reconstituting type I NKT cells as well as selectively blocking or activating type II NKT cells with antibody or the agonist sulfatide, respectively. In this manner, we showed that blockade of both type II NKT cells and Tregs is necessary to abrogate suppression of tumor immunity, but a third cell, the type I NKT cell, determines the balance between these regulatory mechanisms. As patients with cancer often have deficient type I NKT cell function, managing this delicate balance among 3 T-cell subsets may be critical for the success of immunotherapy for human cancer.


Subject(s)
Colorectal Neoplasms/immunology , Kidney Neoplasms/immunology , Natural Killer T-Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Sulfoglycosphingolipids/pharmacology , T-Lymphocyte Subsets/immunology
20.
Proc Natl Acad Sci U S A ; 109(42): 16992-7, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027937

ABSTRACT

To develop more effective vaccines and strategies to regulate chronic inflammatory diseases, it is important to understand the mechanisms of immunological memory. Factors regulating memory CD4(+) T helper (Th)-cell pool size and function remain unclear, however. We show that activation of type I invariant natural killer T (iNKT) cells with glycolipid ligands and activation of type II natural killer T (NKT) cells with the endogenous ligand sulfatide induced dramatic proliferation and expansion of memory, but not naïve, CD4 T cells. NKT cell-induced proliferation of memory Th1 and Th2 cells was dependent largely on the production of IL-2, with Th2-cell proliferation also affected by loss of IL-4. Type II NKT cells were also required for efficient maintenance of memory CD4 T cells in vivo. Activation of iNKT cells resulted in up-regulation of IFN-γ expression by memory Th2 cells. These IFN-γ-producing memory Th2 cells showed a decreased capability to induce Th2 cytokines and eosinophilic airway inflammation. Thus, activated NKT cells directly regulate memory CD4 T-cell pool size and function via the production of cytokines in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Immunologic Memory/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Animals , Antigens, CD1d/genetics , Glycolipids/pharmacology , Immunologic Memory/drug effects , Interferon-gamma/immunology , Interleukin-2/immunology , Interleukin-4/immunology , Mice , Mice, Knockout , Sulfoglycosphingolipids/pharmacology , Th2 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...